Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 39(23): 4461-4474, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-30940718

RESUMO

Excitatory synapses onto somatostatin (SOM) interneurons show robust short-term facilitation. This hallmark feature of SOM interneurons arises from a low initial release probability that regulates the recruitment of interneurons in response to trains of action potentials. Previous work has shown that Elfn1 (extracellular leucine rich repeat and fibronectin Type III domain containing 1) is necessary to generate facilitating synapses onto SOM neurons by recruitment of two separate presynaptic components: mGluR7 (metabotropic glutamate receptor 7) and GluK2-KARs (kainate receptors containing glutamate receptor, ionotropic, kainate 2). Here, we identify how a transsynaptic interaction between Elfn1 and mGluR7 constitutively reduces initial release probability onto mouse cortical SOM neurons. Elfn1 produces glutamate-independent activation of mGluR7 via presynaptic clustering, resulting in a divergence from the canonical "autoreceptor" role of Type III mGluRs, and substantially altering synaptic pharmacology. This structurally induced determination of initial release probability is present at both layer 2/3 and layer 5 synapses. In layer 2/3 SOM neurons, synaptic facilitation in response to spike trains is also dependent on presynaptic GluK2-KARs. In contrast, layer 5 SOM neurons do not exhibit presynaptic GluK2-KAR activity at baseline and show reduced facilitation. GluK2-KAR engagement at synapses onto layer 5 SOM neurons can be induced by calmodulin activation, suggesting that synaptic function can be dynamically regulated. Thus, synaptic facilitation onto SOM interneurons is mediated both by constitutive mGluR7 recruitment by Elfn1 and regulated GluK2-KAR recruitment, which determines the extent of interneuron recruitment in different cortical layers.SIGNIFICANCE STATEMENT This study identifies a novel mechanism for generating constitutive GPCR activity through a transsynaptic Elfn1/mGluR7 structural interaction. The resulting tonic suppression of synaptic release probability deviates from canonical autoreceptor function. Constitutive suppression delays the activation of somatostatin interneurons in circuits, necessitating high-frequency activity for somatostatin interneuron recruitment. Furthermore, variations in the synaptic proteome generate layer-specific differences in facilitation at pyr → SOM synapses. The presence of GluK2 kainate receptors in L2/3 enhances synaptic transmission during prolonged activity. Thus, layer-specific synaptic properties onto somatostatin interneurons are mediated by both constitutive mGluR7 recruitment and regulated GluK2 kainate receptor recruitment, revealing a mechanism that generates diversity in physiological responses of interneurons.


Assuntos
Interneurônios/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de Glutamato Metabotrópico/agonistas , Córtex Somatossensorial/citologia , Somatostatina/análise , Transmissão Sináptica/fisiologia , Regulação Alostérica , Animais , Genes Reporter , Hipocampo/citologia , Interneurônios/química , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Fosfosserina/farmacologia , Propionatos/farmacologia , Receptores de Ácido Caínico/metabolismo , Proteínas Recombinantes/metabolismo , Córtex Somatossensorial/ultraestrutura , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Receptor de GluK2 Cainato
2.
Biochem Biophys Res Commun ; 514(4): 1058-1065, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31097221

RESUMO

Resolvins (Rvs) are endogenous lipid mediators that promote resolution of inflammation and return to homeostasis. We previously reported that RvD1 both facilitates M2 macrophage polarization of Kupffer cells (KCs) and efferocytosis and modulates thioredoxin 2-mediated mitochondrial quality control in liver ischemia/reperfusion (IR) injury. However, the specific cellular or molecular targets of RvD1 remain poorly understood. Sphingosine-1-phosphate (S1P), the natural sphingolipid ligand for a family of G protein-coupled receptors (S1P1-S1P5), regulates lymphocyte circulation and various immune responses. Here we investigated the role of RvD1 in IR-induced hepatocellular damage with a focus on S1P signaling. Male C57BL/6 mice were subjected to partial hepatic ischemia for 60 min, followed by reperfusion. Mice were pretreated with RvD1 (15 µg/kg, i.p.) 1 h prior to ischemia and immediately before reperfusion. To deplete KCs, liposome clodronate was administered (100 µL/mice, i.v.) 24 h prior to ischemia. Mice were pretreated with VPC23019 (100 µg/kg, i.p.), an antagonist for S1P1/S1P3 10 min prior to initial RvD1 treatment. Exogenous RvD1 attenuated IR-induced hepatocellular damage as evidenced by serum HMGB1 release. RvD1 attenuated the decrease in hepatic S1P concentration induced by IR. KC depletion by liposome clodronate did not alter the effect of RvD1 on sphingosine kinases (SKs) and S1P receptors, suggesting independency of KCs. Moreover, in purified hepatocytes of mice exposed to IR, mRNA expression of SK1, SK2, S1P1, and S1P3 decreased significantly, and this was attenuated by RvD1. Finally, VPC23019 pretreatment abolished the hepatoprotective effects of RvD1 in serum HMGB1 release. Our findings suggest that RvD1 protects the liver against IR injury by activating S1P signaling.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Fígado/efeitos dos fármacos , Lisofosfolipídeos/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Esfingosina/análogos & derivados , Animais , Raios Infravermelhos , Fígado/metabolismo , Fígado/patologia , Lisofosfolipídeos/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/antagonistas & inibidores , Esfingosina/metabolismo
3.
Acta Biochim Biophys Sin (Shanghai) ; 51(4): 402-410, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30877755

RESUMO

Epicardial progenitor cells (EpiCs) which are derived from the proepicardium have the potential to differentiate into coronary vascular smooth muscle cells during development. Whether sphingosine 1-phosphate (S1P), a highly hydrophobic zwitterionic lysophospholipid in signal transduction, induces the differentiation of EpiCs is unknown. In the present study, we demonstrated that S1P significantly induced the expression of smooth muscle cell specific markers α-smooth muscle actin and myosin heavy chain 11 in the EpiCs. And the smooth muscle cells differentiated from the EpiCs stimulated by S1P were further evaluated by gel contraction assay. To further confirm the major subtype of sphingosine 1-phosphate receptors (S1PRs) involved in the differentiation of EpiCs, we used the agonists and antagonists of different S1PRs. The results showed that the S1P1/S1P3 antagonist VPC23019 and the S1P2 antagonist JTE013 significantly attenuated EpiCs differentiation, while the S1P1 agonist SEW2871 and antagonist W146 did not affect EpiCs differentiation. These results collectively suggested that S1P, principally through its receptor S1P3, increases EpiCs differentiation into VSMCs and thus indicated the importance of S1P signaling in the embryonic coronary vasculature, while S1P2 plays a secondary role.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Células-Tronco Embrionárias Murinas/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Pericárdio/citologia , Esfingosina/análogos & derivados , Actinas/genética , Actinas/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Pericárdio/embriologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Esfingosina/farmacologia
4.
J Neurochem ; 140(5): 776-786, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28054340

RESUMO

Fingolimod, a sphingosine-1-phosphate receptor (S1PR) agonist, is clinically available to treat multiple sclerosis and is showing promise in treating stroke. We investigated if fingolimod provides long-term protection from experimental neonatal germinal matrix hemorrhage (GMH), aiming to support a potential mechanism of acute fingolimod-induced protection. GMH was induced in P7 rats by infusion of collagenase (0.3 U) into the right ganglionic eminence. Animals killed at 4 weeks post-GMH received low- or high-dose fingolimod (0.25 or 1.0 mg/kg) or vehicle, and underwent neurocognitive testing before histopathological evaluation. Subsequently, a cohort of animals killed at 72 h post-GMH received 1.0 mg/kg fingolimod; the specific S1PR1 agonist, SEW2871; or fingolimod co-administered with the S1PR1/3/4 inhibitor, VPC23019, or the Rac1 inhibitor, EHT1864. All drugs were injected intraperitoneally 1, 24, and 48 h post-surgery. At 72 h post-GMH, brain water content, extravasated Evans blue dye, and hemoglobin were measured as well as the expression levels of phospho-Akt, Akt, GTP-Rac1, Total-Rac1, ZO1, occludin, and claudin-3 determined. Fingolimod significantly improved long-term neurocognitive performance and ameliorated brain tissue loss. At 72 h post-GMH, fingolimod reduced brain water content and Evans blue dye extravasation as well as reversed GMH-induced loss of tight junctional proteins. S1PR1 agonism showed similar protection, whereas S1PR or Rac1 inhibition abolished the protective effect of fingolimod. Fingolimod treatment improved functional and morphological outcomes after GMH, in part, by tempering acute post-hemorrhagic blood-brain barrier disruption via the activation of the S1PR1/Akt/Rac1 pathway.


Assuntos
Cloridrato de Fingolimode/farmacologia , Hemorragias Intracranianas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Água Corporal/metabolismo , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Cognição/efeitos dos fármacos , Feminino , Hemorragias Intracranianas/metabolismo , Hemorragias Intracranianas/psicologia , Contagem de Leucócitos , Masculino , Oxidiazóis/farmacologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Gravidez , Pironas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Tiofenos/farmacologia , Proteínas de Junções Íntimas/metabolismo , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
5.
Biochem Biophys Res Commun ; 476(4): 627-634, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27282481

RESUMO

Endothelial nitric oxide synthase (eNOS) plays a crucial role in vascular homeostasis. Lysophospholipid interaction with sphingosine 1-phosphat (S1P) receptors results in eNOS activation in different cells. In endothelial cells, eNOS activation via S1P1 or S1P3 was shown controversially. The aim of this study is to investigate the meaning of both S1P receptors for eNOS activation in human endothelial cells. Therefore, several S1P1 and S1P3 agonists in combination with antagonists and specific RNAi approach were used. eNOS activation was measured in human umbilical vein endothelial cells (HUVEC) via DAF2-DA-based fluorescence microscopy. For investigation of the signaling pathway, agonists/antagonist studies, RNAi approach, Luminex™ multiplex, and Western Blot were used. In HUVEC, both the S1P1 agonist AUY954 as well as the S1P1,3 agonist FTY720P induced eNOS activation in a time- and dose-dependent manner. Other S1P1 agonists activated eNOS to a lesser extent. The AUY954-induced eNOS activation was blocked by the S1P1 antagonist W146, the combination of W146 and the S1P3 antagonist CAY10444 and the S1P1,3 antagonist VPC23019, but not by CAY10444 indicating the meaning of S1P1 for the AUY954-induced eNOS activation. The FTY720P-induced eNOS activation was blocked only by the combination of W146 and CAY10444 and the combined S1P1,3 antagonist VPC23019, but not by W146 or CAY10444 indicating the importance of both S1P1 and S1P3 for FTY720-induced eNOS activation. These results were confirmed using specific siRNA against S1P1 and S1P3. The S1P1,3 activation results in Akt phosphorylation and subsequent activation of eNOS via phosphorylation at serine(1177) and dephosphorylation at threonine(495). Beside former investigations with rather unspecific S1P receptor activation these data show potent selective S1P1 activation by using AUY954 and with selective S1P receptor inhibition evidence was provided that both S1P1 and S1P3 lead to downstream activation of eNOS in HUVEC in the same experimental setting. Inhibition or knockdown of one of these receptor subtypes did not abolish the eNOS activation and subsequent NO production.


Assuntos
Células Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Anilidas/farmacologia , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo III/química , Organofosfatos/farmacologia , Organofosfonatos/farmacologia , Fosforilação , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Tiazolidinas/farmacologia , Tiofenos/farmacologia , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia
6.
Neural Plast ; 2016: 8072156, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28018679

RESUMO

Among sphingosine 1-phosphate receptors (S1PRs) family, S1PR1 has been shown to be the most highly expressed subtype in neural stem cells (NSCs) and plays a crucial role in the migratory property of NSCs. Recent studies suggested that S1PR1 was expressed abundantly in the hippocampus, a specific neurogenic region in rodent brain for endogenous neurogenesis throughout life. However, the potential association between S1PR1 and neurogenesis in hippocampus following traumatic brain injury (TBI) remains unknown. In this study, the changes of hippocampal S1PR1 expression after TBI and their effects on neurogenesis and neurocognitive function were investigated, focusing on particularly the extracellular signal-regulated kinase (Erk) signaling pathway which had been found to regulate multiple properties of NSCs. The results showed that a marked upregulation of S1PR1 occurred with a peak at 7 days after trauma, revealing an enhancement of proliferation and neuronal differentiation of NSCs in hippocampus due to S1PR1 activation. More importantly, it was suggested that mitogen-activated protein kinase-Erk kinase (MEK)/Erk cascade was required for S1PR1-meidated neurogenesis and neurocognitive recovery following TBI. This study lays a preliminary foundation for future research on promoting hippocampal neurogenesis and improving TBI outcome.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Neurogênese/fisiologia , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Lesões Encefálicas Traumáticas/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Neurogênese/efeitos dos fármacos , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Esfingosina-1-Fosfato
7.
J Biol Chem ; 289(43): 29961-74, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25193666

RESUMO

To define the upstream and downstream signaling specificities of metabotropic glutamate receptors (mGluR), we have examined the ability of representative mGluR of group I, II, and III to be activated by endogenous amino acids and catalyze activation of G proteins coupled to phospholipase C (PLC), or activation of G(i/o) proteins coupled to the ion channel TRPC4ß. Fluorescence-based assays have allowed us to observe interactions not previously reported or clearly identified. We have found that the specificity for endogenous amino acids is remarkably stringent. Even at millimolar levels, structurally similar compounds do not elicit significant activation. As reported previously, the clear exception is L-serine-O-phosphate (L-SOP), which strongly activates group III mGluR, especially mGluR4,-6,-8 but not group I or II mGluR. Whereas L-SOP cannot activate mGluR1 or mGluR2, it acts as a weak antagonist for mGluR1 and a potent antagonist for mGluR2, suggesting that co-recognition of L-glutamate and L-SOP arose early in evolution, and was followed later by divergence of group I and group II mGluR versus group III in l-SOP responses. mGluR7 has low affinity and efficacy for activation by both L-glutamate and L-SOP. Molecular docking studies suggested that residue 74 corresponding to lysine in mGluR4 and asparagine in mGluR7 might play a key role, and, indeed, mutagenesis experiments demonstrated that mutating this residue to lysine in mGluR7 enhances the potency of L-SOP. Experiments with pertussis toxin and dominant-negative Gα(i/o) proteins revealed that mGluR1 couples strongly to TRPC4ß through Gα(i/o), in addition to coupling to PLC through Gα(q/11).


Assuntos
Evolução Molecular , Proteínas de Ligação ao GTP/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Fosfolipases Tipo C/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Genes Dominantes , Ácido Glutâmico/farmacologia , Células HEK293 , Humanos , Ligantes , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Fosfosserina/farmacologia , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ratos , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/química , Transdução de Sinais/efeitos dos fármacos
8.
Biopharm Drug Dispos ; 36(2): 115-25, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25377184

RESUMO

The subcutaneous administration of biologics is highly desirable; however, incomplete bioavailability after s.c. administration remains a major challenge. In this work we investigated the effects of excipient dependent hyperosmolarity on lymphatic uptake and plasma exposure of rituximab as a model protein. Using Swiss Webster (SW) mice as the animal model, we compared the effects of NaCl, mannitol and O-phospho-L-serine (OPLS) on the plasma concentration of rituximab over 5 days after s.c. administration. An increase was observed in plasma concentrations in animals administered rituximab in hypertonic buffer solutions, compared with isotonic buffer. Bioavailability, as estimated by our pharmacokinetic model, increased from 29% in isotonic buffer to 54% in hypertonic buffer containing NaCl, to almost complete bioavailability in hypertonic buffers containing high dose OPLS or mannitol. This improvement in plasma exposure is due to the improved lymphatic trafficking as evident from the increase in the fraction of dose trafficked through the lymph nodes in the presence of hypertonic buffers. The fraction of the dose trafficked through the lymphatics, as estimated by the model, increased from 0.05% in isotonic buffer to 13% in hypertonic buffer containing NaCl to about 30% for hypertonic buffers containing high dose OPLS and mannitol. The data suggest that hypertonic solutions may be a viable option for improving s.c. bioavailability.


Assuntos
Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/farmacocinética , Soluções Hipertônicas/farmacologia , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Animais , Anticorpos Monoclonais Murinos/sangue , Disponibilidade Biológica , Soluções Tampão , Injeções Subcutâneas , Masculino , Manitol/farmacologia , Camundongos , Fosfosserina/farmacologia , Rituximab , Cloreto de Sódio/farmacologia , Trometamina/farmacologia
9.
Cell Biol Int ; 38(3): 309-17, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24155130

RESUMO

Phosphoserine has potential effectiveness as a simple substrate in preparing bone replacement materials, which could enhance bone forming ability. However, there is a need to investigate the independent effect of phosphoserine on osteogenic differentiation of human adipose stem cells (hADSCs). hADSCs were cultured in an osteogenic medium with phosphoserine. Cell proliferation was analysed by CCK8 and osteogenic differentiation was measured by alkaline phosphatase (ALP) activity, von Kossa staining and real time-polymerase chain reaction (RT-PCR). No stimulatory effect of phosphoserine on cell proliferation was noted at Days 1, 4 and 7. Deposition of calcium increased after the addition of phosphoserine. mRNA expression of type I collagen (COL-I), alkaline phosphatase (ALP), osteocalcin (OCN), Osterix, bone morphogenetic protein-2 (BMP-2) and RUNX2 increased markedly with phosphoserine treatment. The BMP-2 antagonist, noggin, and its receptor kinase inhibitors, dorsomorphin and LDN-193189, attenuated phosphoserine-promoted ALP activity. BMP-responsive and Runx2-responsive reporters were activated by phosphoserine treatment. Thus phosphoserine can promote osteogenic differentiation of hADSCs, probably by activating BMP and Runx2 pathways, which could be a promising approach for enhancing osteogenic capacity of cell-based construction in bone tissue engineering.


Assuntos
Adipócitos/efeitos dos fármacos , Proteína Morfogenética Óssea 2/metabolismo , Osteogênese/efeitos dos fármacos , Fosfosserina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Adipócitos/citologia , Adulto , Fosfatase Alcalina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Humanos , Masculino , Osteocalcina/metabolismo , Células Estromais/citologia
10.
Clin Oral Implants Res ; 25(2): e133-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23210635

RESUMO

OBJECTIVES: Phosphoserine-based functionalization has been proposed as a tool to improve integration of endosseous implants by promoting osteoblast adhesion and differentiation in vitro. The present work investigates whether phosphoserine-tethered poly(epsilon-lysine) dendrons, when applied as a film to titanium surfaces, enhance the differentiation of osteoblastic cells and the activation of Wnt/ß-catenin signaling. MATERIALS AND METHODS: These films were tested in a murine model of calvaria-derived MC3T3 osteoblastic cells, primary bone marrow cells and mesenchymal, undifferentiated C2C12 cells. Gene expression was assayed by Real Time PCR, and activation of Wnt signaling pathway was measured with a reporter assay. RESULTS: Dendrons increased expression of alkaline phosphatase and osteocalcin, two osteoblastic markers, in both murine osteoblastic MC3T3 cells and primary bone marrow cells. The expression of osteoprotegerin, a protein opposing osteoclastogenesis was also significantly higher in cells growing on dendron-coated substrates both at 3 and 6 days of culture. Similarly, the mRNA levels of Wisp-2 and of ß-catenin, two Wnt target genes, were also markedly increased in this group at day 6. The activation of this signaling pathway in cells growing on the dendron-coated surfaces was confirmed by use of a TCF/ß-catenin reporter system in the C2C12 cell line. CONCLUSIONS: The findings of the present study show that phosphoserine-tethered poly(epsilon-lysine) dendron films act as stimuli for the activation of specific signal cascades and promote the differentiation of adhering progenitor cells into an osteoblastic phenotype.


Assuntos
Biomimética , Materiais Revestidos Biocompatíveis , Dendrímeros/farmacologia , Lisina/farmacologia , Osteoblastos/fisiologia , Fosfosserina/farmacologia , Titânio/farmacologia , Via de Sinalização Wnt/fisiologia , Condicionamento Ácido do Dente , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores/metabolismo , Adesão Celular , Diferenciação Celular , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Espectrometria de Massas , Camundongos , Microscopia Eletrônica de Varredura , Osteocalcina/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Propriedades de Superfície
11.
Toxicol In Vitro ; 89: 105564, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36736710

RESUMO

Epidemiological studies indicate that human and animal exposure to environmental mercury (Hg) disrupts normal immune system function, but the molecular mechanism responsible for this is still unresolved. We have previously utilized phospho-proteomic mass spectrometry to demonstrate that in the absence of B Cell Receptor (BCR) stimulation, exposure of B cells to Hg induces significant changes to a great many elements of the BCR signaling pathway in a concentration dependent manner. In this report, we have extended those initial findings by utilizing mass spectrometry to evaluate in detail the effect of low-level Hg exposure on BCR induced phospho-proteomic changes. Specifically, murine WEHI-231 B lymphoma cells were exposed to environmentally relevant levels of Hg with or without concomitant BCR stimulation. The cellular phospho-proteomes were then profiled by LC-MS/MS. We found that for low-level exposures, Hg interference with signal transduction across the BCR pathway was predominantly associated with modification of phosphorylation of 12 phosphosites located on seven different proteins. Nine sites were serine, two sites tyrosine and one site threonine. Most of these sites are novel, in the sense that only the two tyrosine and one of the serine sites have previously been reported to be associated with BCR signaling.


Assuntos
Mercúrio , Animais , Camundongos , Humanos , Fosfosserina/metabolismo , Fosfosserina/farmacologia , Mercúrio/toxicidade , Cromatografia Líquida , Proteômica , Linhagem Celular , Espectrometria de Massas em Tandem , Transdução de Sinais , Receptores de Antígenos de Linfócitos B/metabolismo , Proteínas/metabolismo , Fosforilação , Tirosina/metabolismo
12.
J Neurosci Res ; 90(12): 2349-61, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22987212

RESUMO

Glutamate, the major excitatory neurotransmitter in the retina, functions by activation of both ionotropic (iGluR) and metabotropic (mGluR) glutamate receptors. Group III mGluRs, except for mGluR6, are mostly found in the inner plexiform layer (IPL), and their retinal functions are not well known. Therefore, we decided to investigate the effect of mGluRIII on glutamate release and GABAergic amacrine cells in the chick retina. The nonselective mGluRIII agonist L-SOP promoted a decrease in the number of γ-aminobutyric acid (GABA)-positive cells and in the GABA immunoreactivity in all sublayers of the IPL. This effect was prevented by the antagonist MAP-4, by GAT-1 inhibitor, and by antagonists of iGluR. Under the conditions used, L-SOP did not alter endogenous glutamate release. VU0155041, an mGluR4-positive allosteric modulator, reduced GABA immunoreactivity in amacrine cells and in sublayers 2 and 4 of the IPL but evoked an increase in the glutamate released. VU0155041's effect was inhibited by the absence of calcium. AMN082, a selective mGluR7-positive allosteric modulator, also decreased GABA immunoreactivity in amacrine cells and sublayers 1, 2, and 3 and increased glutamate release, and this effect was also inhibited by calcium absence. DCPG, an mGluR8-selective agonist, did not significantly alter GABA immunoreactivity in amacrine cells or glutamate release. However, it did significantly increase GABA immunoreactivity in sublayers 4 and 5. The results suggest that mGluRIIIs are involved in the modulation of glutamate and GABA release in the retina, possibly participating in distinct visual pathways: mGluR4 might be involved with cholinergic circuitry, whereas mGluR7 and mGluR8 might participate, respectively, in the OFF and the ON pathways.


Assuntos
Células Amácrinas/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Receptores de Glutamato Metabotrópico/fisiologia , Ácido gama-Aminobutírico/metabolismo , Células Amácrinas/química , Células Amácrinas/metabolismo , Anilidas/farmacologia , Animais , Compostos Benzidrílicos/farmacologia , Cálcio/fisiologia , Galinhas , Ácidos Cicloexanocarboxílicos/farmacologia , Maleato de Dizocilpina/farmacologia , Proteínas da Membrana Plasmática de Transporte de GABA/fisiologia , Neurônios GABAérgicos/química , Neurônios GABAérgicos/metabolismo , Ácidos Nipecóticos/farmacologia , Oximas/farmacologia , Fosfosserina/farmacologia , Quinoxalinas/farmacologia , Receptores de Glutamato Metabotrópico/agonistas , Ácido gama-Aminobutírico/análise
13.
J Neurosci Res ; 90(5): 1055-66, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22252898

RESUMO

Perfusion of 4-aminopyridine (4-AP) by microdialysis in the hippocampus produces intense epileptiform behavioral and electrical activity and neurodegeneration, resulting from a stimulated release of glutamate from nerve endings. In contrast, accumulation of extracellular glutamate by blockade of its transport in vivo in anesthetized rats is innocuous, and studies in vitro in brain slices suggest that under these conditions glutamate may activate presynaptic group III metabotropic glutamate receptors (mGluRs) and inhibit its own release. Therefore, using microdialysis, EEG recording, and histological evaluation, we studied the effect of increased endogenous extracellular glutamate by blockade of its transport with pyrrolidine dicarboxylic acid (PDC) on the excitotoxic action of 4-AP in the hippocampus of awake rats. We found that up to a 20-fold increase in extracellular glutamate during >90 min with PDC does not induce any sign of excitotoxicity. On the contrary, this glutamate increase notably protected against the 4-AP-induced seizures and neurodegeneration, and, remarkably, this protection was dependent on the time of perfusion with PDC and thus on the duration of extracellular glutamate accumulation. To test whether this protective action was mediated by the activation of group III mGluRs, we used specific antagonists of these receptors and found that they clearly prevented the protective effect of PDC, without affecting the accumulation of extracellular glutamate. We conclude that the spillover of the excess extracellular glutamate activates presynaptic group III mGluRs and inhibits the stimulatory effect of 4-AP on its release, thus preventing the activation of postsynaptic N-methyl-D-aspartate receptors and its deleterious consequences.


Assuntos
Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , 4-Aminopiridina/farmacologia , Aminoácidos/metabolismo , Animais , Ácidos Carboxílicos/farmacologia , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Eletroquímica , Eletroencefalografia , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Antagonistas de Aminoácidos Excitatórios/farmacologia , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Masculino , Microdiálise , Fosfosserina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Piridinas/farmacologia , Ratos , Ratos Wistar , Fatores de Tempo
14.
Cancer Commun (Lond) ; 42(3): 205-222, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35191227

RESUMO

BACKGROUND: Understanding how the tumor microenvironment is shaped by various factors is important for the development of new therapeutic strategies. Tumor cells often undergo spontaneous apoptotic cell death in tumor microenvironment, these apoptotic cells are histologically co-localized with immunosuppressive macrophages. However, the mechanism by which tumor cell apoptosis modulates macrophage polarization is not fully understood. In this study, we aimed to explore the tumor promoting effects of apoptotic tumor cells and the signal pathways involved. METHODS: Apoptotic cells and macrophages in tumors were detected by immunohistochemical staining. Morphological analysis was performed with Giemsa staining. Lipids generated from apoptotic cells were detected by liquid chromatography-mass spectrometry. Phosphatidylserine-containing liposomes were prepared to mimic apoptotic cells. The expression of protein was determined by real-time PCR, immunohistochemistry enzyme-linked immunosorbent assay and Western blotting. Mouse malignant ascites and subcutaneous tumor models were designed for in vivo analysis. Transgenic mice with specific genes knocked out and inhibitors specific to certain proteins were used for the mechanistic studies. RESULTS: The location and the number of apoptotic cells were correlated with that of macrophages in several types of carcinomas. Phosphatidylserine, a lipid molecule generated in apoptotic cells, induced polarization and accumulation of M2-like macrophages in vivo and in vitro. Moreover, sustained administration of phosphoserine promoted tumor growth in the malignant ascites and subcutaneous tumor models. Further analyses suggested that phosphoserine induced a M2-like phenotype in macrophages, which was related to the activation of phosphoserine receptors including T-cell immunoglobin mucin 4 (TIM4) and the FAK-SRC-STAT3 signaling pathway as well as elevated the expression of the histone demethylase Jumonji domain-containing protein 3 (JMJD3). Administration of specific inhibitors of these pathways could reduce tumor progression. CONCLUSIONS: This study suggest that apoptotic cell-generated phosphoserine might be a notable signal for immunosuppressive macrophages in tumors, and the related pathways might be potential therapeutic targets for cancer therapy.


Assuntos
Neoplasias , Fosfatidilserinas , Animais , Apoptose , Ascite/metabolismo , Histona Desmetilases com o Domínio Jumonji , Macrófagos/metabolismo , Camundongos , Neoplasias/metabolismo , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacologia , Fosfosserina/metabolismo , Fosfosserina/farmacologia , Fator de Transcrição STAT3/metabolismo , Microambiente Tumoral
15.
J Neurosci ; 30(31): 10422-30, 2010 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-20685984

RESUMO

Short-term habituation is a basic form of learning that is analyzed in different species and using different behavioral models. Previous studies on mechanisms of short-term habituation yielded evidence for a potential role of group III metabotropic glutamate receptors (mGluRIIIs). Here we tested the hypothesis that mGluRIII mediate short-term habituation of startle in rats, combining electrophysiological experiments in vitro with behavioral studies in vivo. We applied different mGluRIII agonists and antagonists on rat brainstem slices while recording from startle-mediating neurons in the caudal pontine reticular nucleus (PnC) and monitoring synaptic depression presumably underlying habituation. Furthermore, we injected the mGluRIII antagonist (RS)-alpha-phosphonophenylglycine (MPPG) and the agonist L-(+)-2-amino-4-phosphonobutyric acid (L-AP4) into the PnC of rats in vivo and measured its effect on startle habituation. Our results show that activation of mGluRIIIs in the PnC strongly inhibits startle-mediating giant neurons in vitro. Accordingly, L-AP4 reduced startle responses in vivo. However, synaptic depression in the slice was not disrupted by mGluRIII antagonists or agonists. Correspondingly, the in vivo application of the mGluRIII antagonist MPPG failed to show any effect on short-term habituation of startle responses. We therefore conclude that mGluRs are expressed within the primary startle pathway and that they inhibit startle responses upon activation; however, this inhibition does not play any role in synaptic depression and short-term habituation of startle. This is in contrast to the role of mGluRIIIs in other forms of habituation and supports the notion that there are different mechanisms involved in habituation of sensory-evoked behaviors.


Assuntos
Habituação Psicofisiológica/fisiologia , Depressão Sináptica de Longo Prazo/fisiologia , Neurônios/fisiologia , Ponte/fisiologia , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Reflexo de Sobressalto/fisiologia , Formação Reticular/fisiologia , Alanina/análogos & derivados , Alanina/farmacologia , Análise de Variância , Animais , Eletrofisiologia , Potenciais Evocados Auditivos/efeitos dos fármacos , Potenciais Evocados Auditivos/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Habituação Psicofisiológica/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Fosfosserina/farmacologia , Ponte/efeitos dos fármacos , Propionatos/farmacologia , Ratos , Ratos Sprague-Dawley , Formação Reticular/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
16.
Hippocampus ; 21(12): 1302-17, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20824730

RESUMO

Stratum lacunosum-moleculare interneurons (L-Mi) in hippocampal area CA3 target the apical dendrite of pyramidal cells providing feedforward inhibition. Here we report that selective activation of group III metabotropic glutamate receptors (mGluRs) 4/8 with L(+)-2-amino-4-phosphnobytyric acid (L-AP4; 10 µM) decreased the probability of glutamate release from the mossy fiber (MF) terminals synapsing onto L-Mi. Consistent with this interpretation, application of L-AP4 in the presence of 3 mM strontium decreased the frequency of asynchronous MF EPSCs in L-Mi. Furthermore, the dose response curve showed that L-AP4 at 400 µM produced no further decrease in MF EPSC amplitude compared with 20 µM L-AP4, indicating the lack of mGluRs 7 at these MF terminals. We also found that one mechanism of mGluRs 4/8-mediated inhibition of release is linked to N-type voltage gated calcium channels at MF terminals. Application of the group III mGluR antagonist MSOP (100 µM) demonstrated that mGluRs 4/8 are neither tonically active nor activated by low and moderate frequencies of activity. However, trains of stimuli to the MF at 20 and 40 Hz delivered during the application of MSOP revealed a relief of inhibition of transmitter release and an increase in the overall probability of action potential firing in the postsynaptic L-Mi. Interestingly, the time to first action potential was significantly shorter in the presence of MSOP, indicating that mGluR 4/8 activation delays L-Mi firing in response to MF activity. Taken together, our data demonstrate that the timing and probability of action potentials in L-Mi evoked by MF synaptic input is regulated by the activation of presynaptic high affinity group III mGluRs.


Assuntos
Região CA3 Hipocampal/fisiologia , Interneurônios/fisiologia , Fibras Musgosas Hipocampais/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Receptores Pré-Sinápticos/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Aminobutiratos/farmacologia , Animais , Região CA3 Hipocampal/citologia , Região CA3 Hipocampal/efeitos dos fármacos , Ciclopropanos/farmacologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Glicina/análogos & derivados , Glicina/farmacologia , Interneurônios/efeitos dos fármacos , Masculino , Fibras Musgosas Hipocampais/efeitos dos fármacos , Técnicas de Patch-Clamp , Fosfosserina/farmacologia , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Receptores Pré-Sinápticos/efeitos dos fármacos
17.
Anesthesiology ; 114(2): 412-23, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21245732

RESUMO

BACKGROUND: Spinal cord glutamate transporters clear synaptically released glutamate and maintain normal sensory transmission. However, their ultrastructural localization is unknown. Moreover, whether and how they participate in inflammatory pain has not been carefully studied. METHODS: Immunogold labeling with electron microscopy was carried out to characterize synaptic and nonsynaptic localization of glutamate transporters in the superficial dorsal horn. Their expression and uptake activity after formalin- and complete Freund's adjuvant (CFA)-induced inflammation were evaluated by Western blot analysis and glutamate uptake assay. Effects of intrathecal glutamate transporter activator (R)-(-)-5-methyl-1-nicotinoyl-2-pyrazoline and inhibitors (DL-threo-ß-benzyloxyaspartate [TBOA], dihydrokainate, and DL-threo-ß-hydroxyaspartate), or TBOA plus group III metabotropic glutamate receptor antagonist (RS)-α-methylserine-O-phosphate, on formalin- and CFA-induced inflammatory pain were examined. RESULTS: In the superficial dorsal horn, excitatory amino acid carrier 1 is localized in presynaptic membrane, postsynaptic membrane, and axonal and dendritic membranes at nonsynaptic sites, whereas glutamate transporter-1 and glutamate/aspartate transporter are prominent in glial membranes. Although expression of these three spinal glutamate transporters was not altered 1 h after formalin injection or 6 h after CFA injection, glutamate uptake activity was decreased at these time points. Intrathecal (R)-(-)-5-methyl-1-nicotinoyl-2-pyrazoline had no effect on formalin-induced pain behaviors. In contrast, intrathecal TBOA, dihydrokainate, and DL-threo-ß-hydroxyaspartate reduced formalin-evoked pain behaviors in the second phase. Intrathecal TBOA also attenuated CFA-induced thermal hyperalgesia at 6 h after CFA injection. The antinociceptive effects of TBOA were blocked by coadministration of (RS)-α-methylserine-O-phosphate. CONCLUSION: Our findings suggest that spinal glutamate transporter inhibition relieves inflammatory pain through activation of inhibitory presynaptic group III metabotropic glutamate receptors.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Inflamação/metabolismo , Dor/metabolismo , Animais , Ácido Aspártico/farmacologia , Western Blotting , Modelos Animais de Doenças , Formaldeído , Adjuvante de Freund , Ácido Glutâmico/metabolismo , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Masculino , Ácidos Nicotínicos/farmacologia , Fosfosserina/farmacologia , Células do Corno Posterior/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo , Medula Espinal/citologia
18.
Exp Eye Res ; 91(5): 601-12, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20696157

RESUMO

The damaged zebrafish retina replaces lost neurons through a regenerative response that initiates with the asymmetric cell division of Müller glia to produce neuronal progenitor cells that proliferate and migrate to the damaged retinal layer, where they differentiate into the lost neuronal cell types. Because Müller glia are known to phagocytose apoptotic retinal cells during development, we tested if Müller glia engulfed apoptotic rod cell bodies in light-damaged retinas. After 24h of constant intense light, damaged retinas revealed both a strong nuclear TUNEL signal in photoreceptors and a weak cytoplasmic TUNEL signal in Müller glia, although Müller glial apoptosis is not observed in the light-damaged retina. Light damage of a rod-specific transgenic reporter line, Tg(XlRho:EGFP)(fl1), resulted in some Müller glia containing both TUNEL signal and EGFP, which indicated that this subset of Müller glia engulfed apoptotic photoreceptor cell bodies. To determine if phagocytosis induced the Müller glial proliferative response in the light-damaged retina, we utilized O-phospho-l-serine (L-SOP), a molecule that mimics the phosphatidylserine head group and partially blocks microglial phagocytosis of apoptotic cells. Intravitreal injection of L-SOP immediately prior to beginning constant intense light treatment: i) did not significantly reduce light-induced photoreceptor cell death, ii) significantly reduced the number of PCNA-positive Müller glia, and iii) significantly reduced the number of cone photoreceptors in the regenerated retina relative to control retinas. Because L-SOP is also a specific group III metabotropic glutamate receptor (mGluR) agonist, we also tested if the more potent specific group III agonist, L-2-amino-4-phosphonobutyrate (L-AP4), the specific group III antagonist (RS)-α-Methylserine-O-phosphate (MSOP) or the specific group I antagonist, L-2-amino-3-phophonopropanoic acid (L-AP3) affected Müller glial proliferation. We found no changes with any of these factors compared to control retinas, revealing that metabotropic glutamate receptors were not necessary in the Müller glia proliferative response. Furthermore, ascl1a and stat3 expression were unaffected in either the L-SOP or MSOP-injected retinas relative to controls, suggesting L-SOP disrupts Müller glia proliferation subsequent to or in parallel with ascl1a and stat3 activation. This implies that at least one signaling mechanism, in addition to the process disrupted by L-SOP, is required to activate Müller glia proliferation in the light-damaged retina.


Assuntos
Proliferação de Células/efeitos dos fármacos , Luz/efeitos adversos , Neuroglia/patologia , Fagocitose/fisiologia , Fosfosserina/análogos & derivados , Lesões Experimentais por Radiação/patologia , Regeneração/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/fisiologia , Animais , Animais Geneticamente Modificados , Apoptose , Técnica Indireta de Fluorescência para Anticorpo , Marcação In Situ das Extremidades Cortadas , Microscopia Confocal , Fagocitose/efeitos dos fármacos , Fosfosserina/farmacologia , Lesões Experimentais por Radiação/metabolismo , Retina/efeitos da radiação , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra
19.
Acta Biomater ; 105: 280-289, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31945507

RESUMO

Calcium phosphate-based bone cements have been widely adopted in both orthopedic and dental applications. Phosphoserine (pSer), which has a natural role in biomineralization, has been identified to possess the functionality to react with calcium phosphate phases, such as tetracalcium phosphate (TTCP) and α-tricalcium phosphate (α-TCP), and form a uniquely adhesive cement. This study investigated the chemical composition and phase evolution of a heterogeneous calcium phosphate (56% TTCP and 15% α-TCP) and pSer cement system with respect to pH. The coordination network of calcium phosphoserine monohydrate was discovered as the predominant crystalline phase of this adhesive apatitic cement system. Furthermore, it was determined that pH has a significant effect on the reaction kinetics of the system, whereby a lower pH tends to accelerate the reaction rate and favor products with lower Ca/P ratios. These findings provide a better understanding of the reaction and products of this adhesive organo-ceramic cement, which can be compositionally tuned for broad applications in the orthopedic and dental spaces. STATEMENT OF SIGNIFICANCE: The application of self-setting calcium phosphate cements (CPCs) in hard tissue regeneration has been a topic of significant research since their introduction to the field 30 years ago. Traditional CPCs, however, are limited by their suboptimal mechanical properties due to their solely inorganic composition. Recently, it was discovered that monomeric phosphoserine (pSer) is capable of serving as a setting reagent for a subset of CPC systems, resulting in an adhesive organo-ceramic composite. Despite its adhesive functionality and biomedical potential, its reaction chemistry and product composition were not well characterized. The present study identifies a calcium phosphoserine coordination network as the primary crystalline phase of this apatitic cement system and further characterizes compositional tunability of the products with respect to pH.


Assuntos
Apatitas/farmacologia , Cimentos Ósseos/farmacologia , Cálcio/farmacologia , Fosfosserina/farmacologia , Cimentos de Resina/farmacologia , Fosfatos de Cálcio/farmacologia , Concentração de Íons de Hidrogênio , Modelos Moleculares , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
20.
J Neurosci ; 28(18): 4777-84, 2008 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-18448654

RESUMO

Neurotransmitter signaling in the mature nervous system is well understood, but the functions of transmitters in the immature nervous system are less clear. Although transmitters released during embryogenesis regulate neuronal proliferation and migration, little is known about their role in regulating early neuronal differentiation. Here, we show that GABA and glutamate drive calcium-dependent embryonic electrical activity that regulates transmitter specification. The number of neurons expressing different transmitters changes when GABA or glutamate signaling is blocked chronically, either using morpholinos to knock down transmitter-synthetic enzymes or applying pharmacological receptor antagonists during a sensitive period of development. We find that calcium spikes are triggered by metabotropic GABA and glutamate receptors, which engage protein kinases A and C. The results reveal a novel role for embryonically expressed neurotransmitters.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Ácido Glutâmico/metabolismo , Receptores de GABA/fisiologia , Receptores de Glutamato/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Antígenos CD57/metabolismo , Cálcio/metabolismo , Colina O-Acetiltransferase/metabolismo , Embrião não Mamífero , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glutamato Descarboxilase/metabolismo , Pesquisa sobre Serviços de Saúde , Larva , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfosserina/farmacologia , Receptores de GABA/efeitos dos fármacos , Receptores de Glutamato/efeitos dos fármacos , Sinapses/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/metabolismo , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA